UniProt ID: B5R1A9
Host systems: Expressed in E. coli, yeast, baculovirus, or mammalian cells .
Formulation: Tris-based buffer with 50% glycerol for stability .
ELISA development: Utilized as an antigen for antibody detection assays .
Vaccine research: Similar recombinant outer membrane proteins (e.g., SefA, OMPs) have been tested in poultry vaccines to reduce Salmonella colonization .
Recombinant AaeX proteins across bacterial species:
Species | Gene Name | Host System | Purity |
---|---|---|---|
Salmonella enteritidis PT4 | aaeX (SEN3199) | E. coli/Mammalian | ≥85% |
Enterobacter sp. | Ent638_3679 | Yeast/Baculovirus | ≥85% |
Escherichia coli | aaeX | Mammalian | ≥85% |
Functional studies: No direct evidence links AaeX to virulence mechanisms or metabolic pathways.
Structural biology: Predicted domains (e.g., DNA-binding motifs) require crystallographic validation .
Diagnostic utility: Potential as a biomarker for Salmonella enteritidis PT4 detection warrants exploration .
KEGG: set:SEN3199
Salmonella enteritidis PT4 Protein AaeX (aaeX) is a small membrane protein consisting of 67 amino acids with the sequence: MSLFPVIVVFGLSFPPIFFELLLS LAIFWLVRRMVPTGIYDFW WPALFNTALYCCLFYLISRLFV . The protein is encoded by the aaeX gene, identified in the Salmonella enteritidis PT4 strain P125109 genome with the UniProt accession number B5R1A9 .
For structural characterization, researchers typically employ:
Circular dichroism spectroscopy to analyze secondary structure elements
Hydrophobicity analysis suggesting a transmembrane topology
Bioinformatic prediction tools to identify conserved domains
The hydrophobic nature of the N-terminal region indicates a probable membrane-anchoring domain, while the C-terminal region contains charged residues that may interact with cytoplasmic or periplasmic components.
The optimal expression system for recombinant AaeX production depends on research objectives. Based on available data:
For membrane proteins like AaeX, E. coli-based expression systems require optimization of detergents for membrane extraction. Regardless of system, recombinant AaeX proteins should achieve ≥85% purity as determined by SDS-PAGE for reliable experimental use .
Functional verification of recombinant AaeX requires:
Immunological confirmation: Western blotting using anti-AaeX antibodies
Membrane localization assays: Subcellular fractionation followed by detection in membrane fractions
Complementation studies: Introducing recombinant AaeX into aaeX-knockout strains to restore wild-type phenotype
Binding assays: If AaeX interacts with specific host factors
Researchers should establish appropriate positive and negative controls, including:
Positive control: Native AaeX from S. enteritidis PT4
Negative control: Unrelated recombinant protein of similar size
Since AaeX is conserved across multiple bacterial species including Salmonella paratyphi B, Salmonella schwarzengrund, and Escherichia coli , comparative functional analysis between homologs can provide additional verification of protein activity.
Salmonella enterica strains have demonstrated promising potential as delivery vehicles for antitumor molecules, with several key considerations for researchers optimizing such systems:
The development of recombinant Salmonella enterica strains requires strategic attenuation through mutations in specific pathways while preserving tumor-targeting capabilities. Effective attenuating mutations include:
Metabolic pathway modifications:
Virulence reduction modifications:
Lipid A structure modifications (msbB mutations)
Type III secretion system alterations
For therapeutic applications targeting cancer, researchers should consider that Salmonella enterica has demonstrated intrinsic antitumor activity in multiple cancer models:
Murine cancer models (sarcomas, leukemia, colon cancer)
Human cancer xenotransplantation models (prostate, breast, osteosarcoma)
When incorporating AaeX into recombinant Salmonella therapeutic systems, researchers should:
Evaluate if AaeX overexpression affects bacterial tropism for tumor tissues
Determine if AaeX can serve as a fusion partner for delivery of therapeutic proteins
Assess whether AaeX modulates host immune responses that might enhance antitumor effects
As a small membrane protein, AaeX presents several technical challenges for structural characterization:
Challenge | Description | Potential Solutions |
---|---|---|
Hydrophobicity | The hydrophobic nature (MSLFPVIVVFGLSFPPIFFELLLS LAIFWLVR) complicates solubilization | Screen multiple detergents (DDM, LMNG, SDS); consider nanodiscs or amphipols for stabilization |
Small size | At 67 amino acids, obtaining sufficient quantities for NMR or X-ray crystallography is difficult | Express as fusion protein with soluble partners (MBP, SUMO); use solid-state NMR for membrane-embedded studies |
Conformational flexibility | Membrane proteins often have dynamic regions | Utilize molecular dynamics simulations to complement experimental data; consider stabilizing mutations |
Expression yield | Low expression common for membrane proteins | Optimize codon usage; test multiple expression systems; consider synthetic approaches for specific domains |
An integrated structural biology approach combining:
This multi-technique strategy may overcome individual method limitations to elucidate AaeX structure-function relationships.
For researchers investigating AaeX interactions with host immune cells, methodological considerations include:
Cell models selection:
Experimental parameters for cell infection studies:
Parameter | Recommendation | Justification |
---|---|---|
Multiplicity of infection | 10-50 bacteria per cell | Ensures detectable internalization without overwhelming host cells |
Infection duration | 0-48 hours | Covers initial entry (0-2h) and longer survival dynamics |
Temperature | 37°C for mammalian; 41°C for avian cells | Physiologically relevant |
Controls | Wild-type S. enteritidis; ΔaaeX mutant; complemented strain | Isolates AaeX-specific effects |
Quantification methods:
Gentamicin protection assay to distinguish intracellular from extracellular bacteria
Confocal microscopy with fluorescently-labeled bacteria and specific subcellular markers
Flow cytometry for high-throughput analysis of host-pathogen interactions
When comparing results across different cell types, researchers should normalize data to account for differences in phagocytic capacity and bacterial replication rates.
A comprehensive experimental approach to determine AaeX's role in virulence should include:
Genetic manipulation strategies:
CRISPR-Cas9 for precise deletion of aaeX
Complementation with wild-type and mutated versions of aaeX
Conditional expression systems to control timing of aaeX expression
In vitro virulence assays:
Adhesion and invasion assays using epithelial cell lines
Survival within professional phagocytes (macrophages)
Biofilm formation capacity
Resistance to antimicrobial peptides and oxidative stress
In vivo infection models:
Murine typhoid fever model
Chicken gastrointestinal colonization model
Competition assays between wild-type and ΔaaeX mutants
Transcriptomic and proteomic analyses:
RNA-seq to identify genes differentially expressed in ΔaaeX mutants
Comparative proteomics of membrane fractions
Phosphoproteomics to identify signaling pathways affected by AaeX
Data interpretation should consider potential compensatory mechanisms that may mask AaeX phenotypes, necessitating careful experimental design with appropriate controls and statistical power calculations.
For comparative analysis of AaeX homologs across bacterial species:
Bioinformatic approaches:
Position-specific scoring matrix (PSSM) searches instead of simple BLAST
Hidden Markov Model (HMM) profiles of AaeX family
Phylogenetic analysis including potential homologs from Salmonella paratyphi B, Salmonella schwarzengrund, Salmonella heidelberg, and Escherichia coli
Synteny analysis to identify conserved genomic contexts
Functional complementation:
Express heterologous AaeX proteins in S. enteritidis ΔaaeX background
Quantify restoration of phenotypes (invasion, survival, etc.)
Create chimeric proteins from different species to map functional domains
Structure-function relationship studies:
Identify conserved amino acid residues across homologs
Site-directed mutagenesis of these residues
Assess impact on protein localization and function
Species | Gene Name | Protein Size | Identity to S. enteritidis AaeX | Notable Differences |
---|---|---|---|---|
Salmonella paratyphi B | SPAB_04194 | ~67 aa | High | Minimal |
Salmonella schwarzengrund | aaeX | ~67 aa | High | Minimal |
Escherichia coli | aaeX/yhcR | ~67 aa | Moderate | Potential functional divergence |
Serratia proteamaculans | Spro_4392 | ~67 aa | Lower | May have distinct function |
This comparative approach can reveal evolutionary conservation patterns that suggest functional importance of specific protein regions.
For researchers developing immunological detection methods for AaeX:
Antibody development strategies:
Target unique epitopes in the C-terminal region (YCCLFYLISRLFV)
Consider synthetic peptide immunization rather than whole protein
Develop both polyclonal and monoclonal antibodies for different applications
Western blot optimization:
SDS-PAGE with tricine gels optimized for small proteins
Transfer conditions: 100V for 1 hour using PVDF membrane (0.2μm pore size)
Blocking: 5% non-fat milk in TBST (minimal cross-reactivity)
Primary antibody dilution: Start at 1:1000 and optimize
Immunofluorescence protocol refinements:
Fixation: 4% paraformaldehyde followed by membrane permeabilization
Primary antibody incubation: 4°C overnight
Secondary antibody: Fluorophore selection based on imaging system
Controls: Include pre-immune serum and peptide competition controls
ELISA development considerations:
These methodologies should be validated using both recombinant AaeX and native protein from Salmonella enteritidis PT4, with appropriate positive and negative controls throughout.
When designing AaeX fusion proteins for functional studies, researchers should consider:
Fusion orientation decisions:
N-terminal fusions may disrupt membrane localization
C-terminal fusions could interfere with potential functional domains
Consider dual-tagging approaches for confirmation of full-length expression
Tag selection criteria:
Tag Type | Advantages | Limitations | Best Applications |
---|---|---|---|
His6 | Small size, minimal interference | Potential nonspecific binding | Purification, not ideal for localization |
FLAG/HA | Small, highly specific antibodies available | May alter protein trafficking | Immunoprecipitation, localization |
GFP/fluorescent proteins | Direct visualization | Large size may disrupt function | Live imaging, trafficking studies |
Split reporter systems | Allows protein-protein interaction studies | Complex design, potential false positives | Interactome studies |
Linker design principles:
Flexible linkers (GGGGS)n for independent domain function
Rigid linkers (EAAAK)n to separate functional domains
Cleavable linkers for tag removal post-purification
Expression vector selection:
Inducible promoters for controlled expression
Strength of promoter matched to solubility of fusion
Consideration of codon optimization for expression host
Researchers should validate that fusion proteins maintain the expected subcellular localization and conduct comparative functional assays between tagged and untagged versions to ensure tag addition does not significantly alter protein function.
For cell-based assays investigating AaeX in host-pathogen interactions:
Cell culture optimization:
For macrophage lines (HD11, MQ-NCSU): Culture in appropriate media supplemented with 5-10% fetal bovine serum
For lymphocyte lines (LSCC-1104-X5, LSCC-RP9, MDCC-MSB-1): Specialized media formulations with growth factors
Cell passage number: Use cells between passages 5-15 for consistent results
Cell density: Seed at 5×10^5 cells/well for 24-well plates
Infection protocol standardization:
Bacterial preparation: Mid-log phase cultures (OD600 0.4-0.6)
Washing steps: 3× PBS to remove media components that may affect infection
Synchronization of infection: Centrifugation at 500×g for 5 minutes
Duration: 1 hour for initial invasion followed by gentamicin treatment
Critical controls:
Heat-killed bacteria (negative control for active invasion)
Inhibitors of cellular processes (cytochalasin D for actin-dependent uptake)
Positive control strains with known invasion capabilities
Mock-infected cells for baseline cellular responses
Quantification methods standardization:
CFU counting: Plating serial dilutions on selective media
Microscopy: Minimum of 100 cells counted across multiple fields
Flow cytometry: Consistent gating strategy for infected vs. uninfected cells
Based on previous studies with Salmonella enteritidis PT4, researchers should note that macrophages typically show higher initial bacterial uptake but better clearance over 48 hours compared to lymphocyte lines . This pattern may be informative when interpreting AaeX-specific effects.
Understanding AaeX's role in different infection phases requires:
Temporal expression analysis:
qRT-PCR analysis of aaeX expression during:
Early attachment and invasion (0-2 hours)
Intracellular survival phase (2-24 hours)
Persistent infection (24-72 hours)
Promoter-reporter fusions to visualize expression in real-time
Environmental regulation studies:
Expression response to pH changes (gastric to intestinal transition)
Nutrient limitation effects (iron, carbon sources)
Oxidative and nitrosative stress conditions
Host antimicrobial peptide exposure
Tissue-specific function analysis:
Intestinal epithelial models
Macrophage infection models
Gallbladder colonization models
Systemic infection models
Since Salmonella enteritidis PT4 shows differential survival patterns in different host cell types , researchers should investigate whether AaeX expression correlates with bacterial persistence in specific cellular niches, potentially indicating adaptation functions for particular host environments.
To evaluate AaeX as a potential antimicrobial target:
Target validation criteria:
Essentiality: Determine if aaeX is essential for virulence or survival
Conservation: Assess presence and conservation across Salmonella strains
Accessibility: Confirm membrane orientation for potential drug binding
Absence in host: Verify no significant homology to human proteins
Small molecule screening approaches:
In silico docking studies against predicted structure
High-throughput screening using bacterial growth inhibition
Targeted library screening of membrane protein inhibitors
Phenotypic screening for virulence attenuation
Alternative targeting strategies:
Immunological targeting with anti-AaeX antibodies
Peptide inhibitors designed to mimic interaction partners
Antisense oligonucleotides to reduce expression
CRISPR-Cas delivery systems targeting aaeX
Resistance development assessment:
Serial passage experiments with sub-inhibitory concentrations
Whole genome sequencing of resistant variants
Fitness cost analysis of resistance mutations
The presence of AaeX homologs across multiple bacterial species suggests evolutionary conservation that might indicate functional importance, potentially making it a valuable target for broad-spectrum approaches.
Structure-function insights into AaeX could enhance therapeutic applications through:
Engineering principles for improved vaccine vectors:
If AaeX affects membrane properties, modifications could alter antigen presentation
Structure-guided mutations might modulate immunogenicity
Fusion with antigenic epitopes at permissive sites identified through structural analysis
Optimization for cancer therapeutics delivery:
Rational design approaches:
Computational modeling to predict effects of mutations
Alanine scanning mutagenesis to identify critical residues
Domain swapping with homologs to create chimeric proteins with novel properties
Expression system refinements:
Research in this direction could benefit from techniques developed for other recombinant protein expression systems, such as the independently controllable expression elements used in viral vector production systems , adapted for bacterial expression contexts.